2,054
Views
66
CrossRef citations to date
0
Altmetric
Review

Acetyl-CoA carboxylase (ACC) as a therapeutic target for metabolic syndrome and recent developments in ACC1/2 inhibitors

, , , , , , & show all
Pages 917-930 | Received 09 Jun 2019, Accepted 16 Aug 2019, Published online: 29 Aug 2019

References

  • World Health Organization [Internet]. Geneva, Switzerland; [cited 2019 June 2]. Available from: http://www.who.int/gho/ncd/risk_factors/overweight/en/
  • Rinella ME. Nonalcoholic fatty liver disease: a systematic review. JAMA. 2015;313(22):2263–2273.
  • Seravalle G, Grassi G. Obesity and hypertension. Pharmacol Res. 2017;122:1–7.
  • Lu Y, Hajifathalian K, Ezzati M, et al. Metabolic mediators of the effects of body-mass index, overweight, and obesity on coronary heart disease and stroke: a pooled analysis of 97 prospective cohorts with 1.8 million participants. Lancet. 2014;383(9921):970–983.
  • Nakano R, Takebe N, Ono M, et al. Involvement of oxidative stress in atherosclerosis development in subjects with sarcopenic obesity. Obes Sci Pract. 2017;3:212–218.
  • Dyck L, Lynch L. Cancer, obesity and immunometabolism-Connecting the dots. Cancer Lett. 2018;417:11–20.
  • Data was obtained from the World Health Organization [Internet]. Geneva, Switzerland; [cited 2019 June 2]. Available from: http://www.who.int/diabetes/global-report/en/
  • Wakil SJ, Stoops JK, Joshi VC, et al. Fatty acid synthesis and its regulation. Annu Rev Biochem. 1983;52:537–579.
  • Munday MR, Hemingway CJ. The regulation of acetyl-CoA carboxylase – a potential target for the action of hypolipidemic agents. Advan Enzyme Regul. 1999;39:205–234.
  • Barber MC, Price NT, Travers MT, et al. Structure and regulation of acetyl-CoA carboxylase genes of metazoa. Biochim Biophys Acta. 2005;1733:1–28.
  • Tong L. Acetyl-coenzyme A carboxylase: crucial metabolic enzyme and attractive target for drug discovery. Cell Mol Life Sci. 2005;62:1784–1803.
  • Abu-Elheiga L, Brinkley WR, Zhong L, et al. The subcellular localization of acetyl-CoA carboxylase 2. Proc Natl Acad Sci USA. 2000;97:1444–1449.
  • Munday MR, Campbell DG, Carling D, et al. Identification by amino acid sequencing of three major regulatory phosphorylation sites on rat acetyl-CoA carboxylase. Eur J Biochem. 1988;175:331–338.
  • Oh SY, Park SK, Kim JW, et al. Acetyl-CoA carboxylase β gene is regulated by sterol regulatory element-binding protein-1 in liver. J Biol Chem. 2003;278:28410–28417.
  • Lin J, Yang R, Tarr PT, et al. Hyperlipidemic effects of dietary saturated fats mediated through PGC-1β coactivation of SREBP. Cell. 2005;120:261–273.
  • Dentin R, Girard J, Postic C, et al. Carbohydrate responsive element binding protein (ChREBP) and sterol regulatory element binding protein-1c (SREBP-1c): two key regulators of glucose metabolism and lipid synthesis in liver. Biochimie. 2005;87:81–86.
  • Kaushik VK, Kavana M, Volz JM, et al. Characterization of recombinant human acetyl-CoA carboxylase-2 steady-state kinetics. Biochim Biophys Acta. 2009;1794:961–967.
  • Cho YS, Lee JI, Shin D, et al. Molecular mechanism for the regulation of human ACC2 through phosphorylation by AMPK. Biochem Biophys Res Commun. 2010;391:187–192.
  • Mao J, DeMayo FJ, Li H, et al. Liver-specific deletion of acetyl-CoA carboxylase 1 reduces hepatic triglyceride accumulation without affecting glucose homeostasis. Proc Natl Acad Sci USA. 2006;103:8552–8557.
  • Mao J, Yang T, Gu Z, et al. aP2-Cre-mediated inactivation of acetyl-CoA carboxylase 1 causes growth retardation and reduced lipid accumulation in adipose tissues. Proc Natl Acad Sci USA. 2009;106:17576–17581.
  • Abu-Elheiga L, Matzuk MM, Kordari P, et al. Mutant mice lacking acetyl-CoA carboxylase 1 are embryonically lethal. Proc Natl Acad Sci USA. 2005;102:12011–12016.
  • Oh W, Abu-Elheiga L, Kordari P, et al. Glucose and fat metabolism in adipose tissue of acetyl-CoA carboxylase 2 knockout mice. Proc Natl Acad Sci USA. 2004;102:1384–1389.
  • Abu-Elheiga L, Matzuk MM, Abo-Hashema KAH. Continuous fatty acid oxidation and reduced fat storage in mice lacking acetyl-coa carboxylase 2. Science. 2001;291:2613–2616.
  • Abu-Elheiga L, Oh W, Kordari P, et al. Acetyl-CoA carboxylase 2 mutant mice are protected against obesity and diabetes induced by high-fat high-carbohydrate diets. Proc Natl Acad Sci USA. 2003;100:10207–10212.
  • Ronnebaum SM, Joseph JW, Ilkayeva O, et al. Chronic suppression of acetyl-CoA carboxylase 1 in beta-cells impairs insulin secretion via inhibition of glucose rather than lipid metabolism. J Biol Chem. 2008;283:14248–14256.
  • Glien M, Haschke G, Schroeter K, et al. Stimulation of fat oxidation, but no sustained reduction of hepatic lipids by prolonged pharmacological inhibition of acetyl CoA carboxylase. Horm Metab Res. 2011;43:601–606.
  • Corbett JW, Harwood J, James H, et al. Inhibitors of mammalian acetyl-CoA carboxylase. Recent Pat Cardiovasc Drug Discov. 2007;2(3):162–180.
  • Gerth K, Bedorf N, Irschik H, et al. The soraphens: A family of novel antifungal compounds froms orangium cellulosum (myxobacteria). J Antibiot (Tokyo). 1993;47:23–31.
  • Vahlensieck HF, Pridzun L, Reichenbach H, et al. Identification of the yeast ACC1 gene product (acetyl-CoA carboxylase) as the target of the polketide fungicide soraphen A. Curr Genet. 1994;25:95–100.
  • Shen Y, Volrath SL, Weatherly SC, et al. A mechanism for the potent inhibition of eukaryotic acetyl-coenzyme a carboxylase by Soraphen A, a macrocyclic polyketide natural product. Mol Cell. 2004;16:881–891.
  • Weatherly SC, Volrath SL, Elich TD, et al. Expression and characterization of recombinant fungal acetyl-CoA carboxylase and isolation of a soraphen-binding domain. Biochem J. 2004;380(Pt 1):105–110.
  • Schreurs M, Van Dijk TH, Gerding A, et al. An inhibitor of the acetyl-CoA carboxylase system, improves peripheral insulin sensitivity in mice fed a high-fat diet. Diabetes Obesity Metab. 2009;11:987–991.
  • Jump DB, Torres-Gonzalez M, Olson LK, et al. Soraphen A, an inhibitor of acetyl CoA carboxylase activity, interferes with fatty acid elongation. Biochem Pharmacol. 2011;81:649–660.
  • Beckers A, Organe S, Timmermans L, et al. Chemical inhibition of acetyl-CoA carboxylase induces growth arrest and cytotoxicity selectively in cancer cells. Cancer Res. 2007;67:8180–8187.
  • Vincent G, Mansfield DJ, Vors JP, et al. Studies toward soraphen A: an aldol-metathesis avenue to the macrocyclic framework. Org Lett. 2006;8(13):2791–2794.
  • Trost BM, Sieber JD, Qian W, et al. Asymmetric total synthesis of soraphen A: A flexible alkyne strategy. Angew Chem Int Ed. 2009;48:5478–5481.
  • Lu HH, Raja A, Franke R, et al. Synthesis and biological evaluation of Paleo-Soraphens. Angew Chem Int Ed. 2013;52:13549–13552.
  • Lu HH, Hinkelmann B, Tautz T, et al. Paleo-Soraphens: chemical total syntheses and biological studies. Org Biomol Chem. 2015 Aug 07;13(29):8029−8036.
  • Canterbury DP, Scott KEN, Kubo O, et al. Synthesis of C11-desmethoxy soraphen A1α: A natural product analogue that inhibits acetyl-CoA carboxylase. ACS Med Chem Lett. 2013;4:1244−1248.
  • Stoiber K, Nagło O, Pernpeintner C, et al. Targeting de novo lipogenesis as a novel approach in anti-cancer therapy. Br J Cancer. 2018;118(1):1–9.
  • Corominas-Faja B, Cuyàs E, Gumuzio J, et al. Chemical inhibition of acetyl-CoA carboxylase suppresses self-renewal growth of cancer stem cells. Oncotarget. 2014;5(18):8306–8316.
  • Rysman E, Brusselmans K, Scheys K, et al. De novo lipogenesis protects cancer cells from free radicals and chemotherapeutics by promoting membrane lipid saturation. Cancer Res. 2010;70(20):8117–8126.
  • Harwood HJ, Petras SF, Shelly LD, et al. Isozyme-nonselective N-substituted bipiperidylcarboxamide acetyl-CoA carboxylase inhibitors reduce tissue malonyl-CoA concentrations, inhibit fatty acid synthesis, and increase fatty acid oxidation in cultured cells and in experimental animals. J Biol Chem. 2003;278:37099–37111.
  • Zhang H, Tweel B, Li J, et al. Crystal structure of the carboxytransferase domain of acetyl-CoA carboxylase in complex with CP-640186. Structure. 2004;12:1683−1691.
  • Madauss KP, Burkhart WA, Consler TG, et al. The human ACC2 CT-domain C-terminus is required for full functionality and has a novel twist. Acta Cryst. 2009;D65:449–461.
  • Zhang H, Yang Z, Shen Y, et al. Crystal structure of the carboxyltransferase domain of acetyl-coenzyme A carboxylase. Science. 2003;299:2064–2067.
  • Chonan T, Oi T, Yamamoto D, et al. (4-Piperidinyl)-piperazine: A new platform for acetyl-CoA carboxylase inhibitors. Bioorg Med Chem Lett. 2009;19:6645–6648.
  • Chonan T, Tanaka H, Yamamoto D, et al. Design and synthesis of disubstituted (4-piperidinyl)-piperazine derivatives as potent acetyl-CoA carboxylase inhibitors. Bioorg Med Chem Lett. 2010;20:3965–3968.
  • Chonan T, Wakasugi D, Yamamoto D, et al. Discovery of novel (4-piperidinyl)-piperazines as potent and orally active acetyl-CoA carboxylase 1/2 non-selective inhibitors: F-Boc and triF-Boc groups are acid-stable bioisosteres for the Boc group. Bioorg Med Chem. 2011;19:1580–1593.
  • Vyas VK, Dabasia M, Qureshi G, et al. Molecular modeling study for the design of novel Acetyl-CoA carboxylase inhibitors using 3D QSAR, molecular docking and dynamic simulations. J Biomol Struct Dyn. 2017;35:2003–2015.
  • Yamashita T, Kamata M, Endo S, et al. Design, synthesis, and structure-activity relationships of spirolactones bearing 2-ureidobenzothiophene as acetyl-CoA carboxylases inhibitors. Bioorg Med Chem Lett. 2011;21:6314–6318.
  • Kamata M, Yamashita T, Kina A, et al. Design, synthesis, and structure–activity relationships of novel spiro-piperidines as acetyl-CoA carboxylase inhibitors. Bioorg Med Chem Lett. 2012;22:3643–3647.
  • Kamata M, Yamashita T, Kina A, et al. Symmetrical approach of spiro-pyrazolidinediones as acetyl-CoAvcarboxylase inhibitors. Bioorg Med Chem Lett. 2012;22:4769–4772.
  • Corbett JW, Freeman-Cook KD, Elliott R, et al. Discovery of small molecule isozyme non-specific inhibitors of mammalian acetyl-CoA carboxylase 1 and 2. Bioorg Med Chem Lett. 2010;20:2383–2388.
  • Freeman-Cook KD, Amor P, Bader S, et al. Maximizing lipophilic efficiency: the use of free-Wilson analysis in the design of inhibitors of acetyl-CoA carboxylase. J Med Chem. 2012;55:935−942.
  • Bagley SW, Southers JA, Cabral S, et al. Synthesis of 7-Oxo-dihydrospiro [indazole-5,4′-piperidine] acetyl-CoA carboxylase inhibitors. J Org Chem. 2012;77:1497−1506.
  • Huard K, Bagley SW, Menhaji-Klotz E, et al. Synthesis of spiropiperidine lactam acetyl-CoA carboxylase inhibitors. J Org Chem. 2012;77:10050−10057.
  • Griffith DA, Dow RL, Huard K, et al. Spirolactam-based Acetyl-CoA Carboxylase inhibitors: toward improved metabolic stability of a chromanone lead structure. J Med Chem. 2013;56:7110−7119.
  • Griffith DA, Kung DW, Esler WP, et al. Decreasing the rate of metabolic ketone reduction in the discovery of a clinical acetyl-CoA carboxylase inhibitor for the treatment of diabetes. J Med Chem. 2014;57:10512–10526.
  • Kim CW, Addy C, Kusunoki J, et al. Acetyl CoA carboxylase inhibition reduces hepatic steatosis but elevates plasma triglycerides in mice and humans: a bedside to bench investigation. Cell Metab. 2017;26:394–406 e6.
  • Bergman A, Gonzalez SC, Tarabar S, et al. Safety, tolerability, pharmacokinetics and pharmacodynamics of a liver-targeting ACC inhibitor (PF-05221304) following single and multiple oral doses. J Hepatol. 2018;68(Suppl 1):S582.
  • Esler W, Ross T, Bergman A, et al. Partial inhibition of de novo lipogenesis with the acetyl-CoA carboxylase inhibitor PF-05221304 does not increase circulating triglycerides in humans and is sufficient to lower steatosis in rats. J Hepatol. 2019;70(1):e69.
  • Parker RA, Kariya T, Grisar JM, et al. 5-(tetradecyloxy)-2-furancarboxylic acid and related hypolipidemic fatty acid-like alkyloxyarylcarboxylic acids. J Med Chem. 1977;20:781–791.
  • Harris RA, McCune SA. 5-(tetradecyloxy)-2-furoic acid. Methods Enzymol. 1981;72:552–559.
  • Hunt DW, Winters GC, Brownsey RW, et al. Inhibition of sebum production with the acetyl coenzyme a carboxylase inhibitor olumacostat glasaretil. J Invest Dermatol. 2017;137:1415–1423.
  • Melnik BC. Olumacostat Glasaretil, a promising topical sebum-suppressing agent that affects all major pathogenic factors of acne vulgaris. J Invest Dermatol. 2017;137:1405–1408.
  • Melnik BC. Linking diet to acne metabolomics, inflammation, and comedogenesis: an update. Clin Cosmet Investig Dermatol. 2015;8:371–388.
  • Bissonnette R, Poulin Y, Drew J, et al. Olumacostat glasaretil, a novel topical sebum inhibitor, in the treatment of acne vulgaris: A phase IIa, multicenter, randomized, vehicle-controlled study. J Am Acad Dermatol. 2017;76:33–39.
  • Zouboulis CC, Dessinioti C, Tsatsou F, et al. Anti-acne drugs in phase 1 and 2 clinical trials. Expert Opin Investig Drugs. 2017;26(7):813–823.
  • Harrimana G, Greenwood J, Bhat S, et al. Acetyl-CoA carboxylase inhibition by ND-630 reduces hepatic steatosis, improves insulin sensitivity, and modulates dyslipidemia in rats. Proc Natl Acad Sci USA. 2016;113:E1796–E1805.
  • Bourbeau MP, Bartberger MD. Recent advances in the development of acetyl-CoA carboxylase (ACC) inhibitors for the treatment of metabolic disease. J Med Chem. 2015;58(2):525–536.
  • Stiede K, Miao W, Blanchette HS, et al. Acetyl-Coenzyme A Carboxylase inhibition reduces de novo lipogenesis in overweight male subjects: a randomized, double-blind, crossover study. Hepatology. 2017;66:324–334.
  • Loomba R, Kayali Z, Noureddin M, et al. GS-0976 reduces hepatic steatosis and fibrosis markers in patients with nonalcoholic fatty liver disease. Gastroenterology. 2018;154.
  • Lawitz EJ, Coste A, Poordad F, et al. Acetyl-CoA carboxylase inhibitor GS-0976 for 12 weeks reduces hepatic de novo lipogenesis and steatosis in patients with nonalcoholic steatohepatitis. Clin Gastroenterol Hepatol. 2018 Apr 26. 16:1983–1991.e3.
  • Harrison S, Noureddin M, Herring R, et al. Preliminary efficacy and safety of acetyl-CoA carboxylase inhibitor GS-0976 in patients with compensated cirrhosis due to NASH. Gastroenterology. 2018;154(6):S–1166.
  • Mantry P, Kayali Z, Noureddin M, et al. Characterization of changes in lipoprotein profiles of patients with nonalcoholic steatohepatitis treated with the acetyl-CoA carboxylase inhibitor GS-0976. J Hepatol. 2018;68:S583–S584.
  • Burton JD, Gronwald JW, Somers DA, et al. Inhibition of plant acetyl-coenzyme a carboxylase by the herbicides sethoxydim and haloxyfop. Biochem Biophys Res Commun. 1987;148(3):1039–1044.
  • Burton JD, Gronwald JW, Keith RA, et al. Kinetics of inhibition of acetyl-coenzyme A carboxylase by sethoxydim and haloxyfop. Pestic Biochem Physiol. 1991;39(2):100–109.
  • Kemal C, Casida JE Coenzyme a esters of 2-aryloxyphenoxypropionate herbicides and 2-arylpropionate antiinflammatory drugs are potent and stereoselective inhibitors of rat liver acetyl-CoA carboxylase. Life Sci. 1992;50(7):533–540.
  • Zhang H, Tweel B, Tong L, et al. Molecular basis for the inhibition of the carboxyltransferase domain of acetyl-coenzyme-A carboxylase by haloxyfop and diclofop. Proc Natl Acad Sci USA. 2004;101(16):5910–5915.
  • Xiang S, Callaghan MM. A different mechanism for the inhibition of the carboxyltransferase domain of acetyl-coenzyme A carboxylase by tepraloxydim. Proc Natl Acad Sci USA. 2009;106(49):20723–20727.
  • Linda PC, Kim YS, Tong L, et al. Mechanism for the inhibition of the carboxyltransferase domain of acetyl-coenzyme A carboxylase by pinoxaden. Proc Natl Acad Sci USA. 2010;107(51):22072–22077.
  • Gu YG, Weitzberg M, Clark RF, et al. Synthesis and structure-activity relationships of N-{3-[2-(4-Alkoxyphenoxy)thiazol-5-yl]-1-methylprop-2-ynyl}carboxy derivatives as selective acetyl-CoA carboxylase 2 inhibitors. J Med Chem. 2006;49:3770–3773.
  • Clark RF, Zhang T, Xin Z, et al. Structure–activity relationships for a novel series of thiazolyl phenyl ether derivatives exhibiting potent and selective acetyl-CoA carboxylase 2 inhibitory activity. Bioorg Med Chem Lett. 2006;16:6078–6081.
  • Gu YG, Weitzberg M, Clark RF, et al. N-{3-[2-(4-Alkoxyphenoxy)thiazol-5-yl]-1-methylprop-2-ynyl}carboxy derivatives as acetyl-CoA carboxylase inhibitors improvement of cardiovascular and neurological liabilities via structural modifications. J Med Chem. 2007;50:1078–1082.
  • Nishiura Y, Matsumura A, Kobayashi N, et al. Discovery of a novel olefin derivative as a highly potent and selective acetyl-CoA carboxylase 2 inhibitor with in vivo efficacy. Bioorg Med Chem Lett. 2018;28:2498–2503.
  • Mizojiri R, Asano M, Tomita D, et al. Discovery of novel selective Acetyl-CoA carboxylase (ACC) 1 inhibitors. J Med Chem. 2018;61:1098−1117.
  • Bengtsson C, Blaho S, Saitton DB, et al. Design of small molecule inhibitors of acetyl-CoA carboxylase 1 and 2 showing reduction of hepatic malonyl-CoA levels in vivo in obese Zucker rats. Bioorg Med Chem. 2011;19:3039–3053.
  • Keil S, Müller M, Zoller G, et al. Identification and synthesis of novel inhibitors of acetyl-CoA carboxylase with in vitro and in vivo efficacy on fat oxidation. J Med Chem. 2010;53:8679–8687.
  • Bourbeau MP, Siegmund A, Allen JG, et al. Piperazine oxadiazole inhibitors of acetyl-CoA carboxylase. J Med Chem. 2013;56:10132−10141.
  • Okazaki S, Noguchi-Yachide T, Sakai T, et al. Discovery of N-(1-(3-(4-phenoxyphenyl)-1,2,4-oxadiazol-5-yl)ethyl) acetamides as novel acetyl-CoA carboxylase 2 (ACC2) inhibitors with peroxisome proliferator-activated receptor α/δ (PPAR α/δ) dual agonistic activity. Bioorg Med Chem. 2016;24:5258–5269.
  • Esler WP, Tesz GJ, Hellerstein MK, et al. Human sebum requires de novo lipogenesis, which is increased in acne vulgaris and suppressed by acetyl-CoA carboxylase inhibition. Sci Transl Med. 2019;11:eaau8465.
  • Zhan Y, Ginanni N, Tota MR, et al. Control of cell growth and survival by enzymes of the fatty acid synthesis pathway in HCT-116 colon cancer cells. Clin Cancer Res. 2008;14:5735–5742.
  • Chajes V, Cambot M, Moreau K, et al. Acetyl-CoA carboxylase alpha is essential to breast cancer cell survival. Cancer Res. 2006;66:5287–5294.
  • Brusselmans K, Schrijver ED, Verhoeven G, et al. RNA interference-mediated silencing of the acetyl-CoA carboxylase-a gene induces growth inhibition and apoptosis of prostate cancer cells. Cancer Res. 2005;65:6719–6725.
  • Petrova E, Scholz A, Paul J, et al. Acetyl-CoA carboxylase inhibitors attenuate WNT and Hedgehog signaling and suppress pancreatic tumor growth. Oncotarget. 2017;8:48660–48670.
  • Jones JEC, Esler WP, Patel R, et al. Inhibition of Acetyl-CoA Carboxylase 1 (ACC1) and 2 (ACC2) reduces proliferation and de novo lipogenesis of EGFRvIII human glioblastoma cells. PLoS ONE. 2017;12(1):e0169566.
  • Saggerson D. Malonyl-CoA, a key signaling molecule in mammalian cells. Annu Rev Nutr. 2008;28:253–272.
  • Harwood HJ. Treating the metabolic syndrome: acetyl-CoA carboxylase inhibition. Expert Opin Ther Targets. 2005;9(2):267–281.
  • Currie E, Schulze A, Zechner R. Cellular fatty acid metabolism and cancer. Cell Metab. 2013;18:153–161.
  • Esler WP, Bence KK. Metabolic targets in nonalcoholic fatty liver disease. Cell Mol Gastroenterol Hepatol. 2019 Apr 18. 8:247–267
  • Kim KH, Lee MS. Pathogenesis of nonalcoholic steatohepatitis and hormone-based therapeutic approaches. Front Endocrinol. 2018;9:485.
  • Yu XX, Murray SF, Pandey SK, et al. Antisense oligonucleotide reduction of DGAT2 expression improves hepatic steatosis and hyperlipidemia in obese mice. Hepatology. 2005;42:362–371.
  • Choi CS, Savage DB, Kulkarni A, et al. Suppression of diacylglycerol acyltransferase-2 (DGAT2), but not DGAT1, with antisense oligonucleotides reverses dietinduced hepatic steatosis and insulin resistance. J Biol Chem. 2007;282:22678–22688.
  • Liu X, Xue R, Ji L, et al. Activation of farnesoid X receptor (FXR) protects against fructose-induced liver steatosis via inflammatory inhibition and ADRP reduction. Biochem Biophys Res Commun. 2014;450(1):117–123.
  • Cariou B. The farnesoid X receptor (FXR) as a new target in non-alcoholic steatohepatitis Le récepteur nucléaire FXR (farnesoid X receptor): une nouvelle cible moléculaire pour le traitement de la NASH. Diabetes Metab. 2008;34(6):685–691.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.